1
|
Kang C, Li X, Yang X, Cheng X, Zhang D, Wei X. Voltage-gated potassium channels associated with head and neck cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189340. [PMID: 40318770 DOI: 10.1016/j.bbcan.2025.189340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 04/28/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
Head and neck cancer (HNC) is a common disease in otorhinolaryngology. Its prevalence is higher in men than in women and is mostly related to tobacco, alcohol and viral infections. Despite significant advances in the treatment of HNC in recent years, the mortality rate is still high and most patients are diagnosed at an advanced stage, and the prognosis for these patients is even worse. Earlier metastasis makes the treatment of HNC trickier. Therefore, actively seeking ways to treat HNC more effectively has been the goal of head and neck surgeons. Potassium (K+) channels are the most diverse ion channels found in all areas of life. Voltage-gated potassium (Kv) channels are the most important subfamily of K+ channels. Multiple Kv channels are associated with the development of HNC. This review focuses on several Kv channels associated with HNC.
Collapse
Affiliation(s)
- Chenglin Kang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China; Department of Otolaryngology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Xiaomei Li
- Department of Otolaryngology, Second People's Hospital of Gansu Province, Lanzhou, Gansu, China
| | - Xiaolong Yang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China; Department of Otolaryngology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Xiaoling Cheng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China; Department of Otolaryngology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Dengxiao Zhang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China; Department of Otolaryngology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Xudong Wei
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China; Department of Otolaryngology, Gansu Provincial Hospital, Lanzhou, Gansu, China.
| |
Collapse
|
2
|
Lill CB, Fitter S, Zannettino ACW, Vandyke K, Noll JE. Molecular and cellular mechanisms of chemoresistance in paediatric pre-B cell acute lymphoblastic leukaemia. Cancer Metastasis Rev 2024; 43:1385-1399. [PMID: 39102101 PMCID: PMC11554931 DOI: 10.1007/s10555-024-10203-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 07/24/2024] [Indexed: 08/06/2024]
Abstract
Paediatric patients with relapsed B cell acute lymphoblastic leukaemia (B-ALL) have poor prognosis, as relapse-causing clones are often refractory to common chemotherapeutics. While the molecular mechanisms leading to chemoresistance are varied, significant evidence suggests interactions between B-ALL blasts and cells within the bone marrow microenvironment modulate chemotherapy sensitivity. Importantly, bone marrow mesenchymal stem cells (BM-MSCs) and BM adipocytes are known to support B-ALL cells through multiple distinct molecular mechanisms. This review discusses the contribution of integrin-mediated B-ALL/BM-MSC signalling and asparagine supplementation in B-ALL chemoresistance. In addition, the role of adipocytes in sequestering anthracyclines and generating a BM niche favourable for B-ALL survival is explored. Furthermore, this review discusses the role of BM-MSCs and adipocytes in promoting a quiescent and chemoresistant B-ALL phenotype. Novel treatments which target these mechanisms are discussed herein, and are needed to improve dismal outcomes in patients with relapsed/refractory disease.
Collapse
Affiliation(s)
- Caleb B Lill
- Myeloma Research Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
- Precision Cancer Medicine Theme, Solid Tumour Program, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Stephen Fitter
- Myeloma Research Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
- Precision Cancer Medicine Theme, Solid Tumour Program, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Andrew C W Zannettino
- Myeloma Research Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
- Precision Cancer Medicine Theme, Solid Tumour Program, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Kate Vandyke
- Myeloma Research Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
- Precision Cancer Medicine Theme, Solid Tumour Program, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Jacqueline E Noll
- Myeloma Research Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia.
- Precision Cancer Medicine Theme, Solid Tumour Program, South Australian Health and Medical Research Institute, Adelaide, Australia.
| |
Collapse
|
3
|
Arcangeli A, Iorio J, Duranti C. Targeting the hERG1 and β1 integrin complex for cancer treatment. Expert Opin Ther Targets 2024; 28:145-157. [PMID: 38372580 DOI: 10.1080/14728222.2024.2318449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 02/09/2024] [Indexed: 02/20/2024]
Abstract
INTRODUCTION Despite great advances, novel therapeutic targets and strategies are still needed, in particular for some carcinomas in the metastatic stage (breast cancer, colorectal cancer, pancreatic ductal adenocarcinoma and the clear cell renal carcinoma). Ion channels may be considered good cancer biomarkers and targets for antineoplastic therapy. These concepts are particularly relevant considering the hERG1 potassium channel as a novel target for antineoplastic therapy. AREAS COVERED A great deal of evidence demonstrates that hERG1 is aberrantly expressed in human cancers, in particular in aggressive carcinomas. A relevant cornerstone was the discovery that, in cancer cells, the channel is present in a very peculiar conformation, strictly bound to the β1 subunit of integrin receptors. The hERG1/β1 integrin complex does not occur in the heart. Starting from this evidence, we developed a novel single chain bispecific antibody (scDb-hERG1-β1), which specifically targets the hERG1/β1 integrin complex and exerts antineoplastic effects in preclinical experiments. EXPERT OPINION Since hERG1 blockade cannot be pursued for antineoplastic therapy due to the severe cardiac toxic effects (ventricular arrhythmias) that many hERG1 blockers exert, different strategies must be identified to specifically target hERG1 in cancer. The targeting of the hERG1/β1 integrin complex through the bispecific antibody scDb-hERG1-β1 can overcome such hindrances.
Collapse
Affiliation(s)
- Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence, Firenze, Italy
- CSDC (Center for the Study of complex dynamics), University of Florence, Sesto Fiorentino (FI), Italy
- MCK Therapeutics srl, Pistoia (PT), Italy
| | - Jessica Iorio
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence, Firenze, Italy
| | - Claudia Duranti
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence, Firenze, Italy
- MCK Therapeutics srl, Pistoia (PT), Italy
| |
Collapse
|
4
|
Capitani C, Chioccioli Altadonna G, Santillo M, Lastraioli E. Ion channels in lung cancer: biological and clinical relevance. Front Pharmacol 2023; 14:1283623. [PMID: 37942486 PMCID: PMC10627838 DOI: 10.3389/fphar.2023.1283623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 10/16/2023] [Indexed: 11/10/2023] Open
Abstract
Despite improvements in treatment, lung cancer is still a major health problem worldwide. Among lung cancer subtypes, the most frequent is represented by adenocarcinoma (belonging to the Non-Small Cell Lung Cancer class) although the most challenging and harder to treat is represented by Small Cell Lung Cancer, that occurs at lower frequency but has the worst prognosis. For these reasons, the standard of care for these patients is represented by a combination of surgery, radiation therapy and chemotherapy. In this view, searching for novel biomarkers that might help both in diagnosis and therapy is mandatory. In the last 30 years it was demonstrated that different families of ion channels are overexpressed in both lung cancer cell lines and primary tumours. The altered ion channel profile may be advantageous for diagnostic and therapeutic purposes since most of them are localised on the plasma membrane thus their detection is quite easy, as well as their block with specific drugs and antibodies. This review focuses on ion channels (Potassium, Sodium, Calcium, Chloride, Anion and Nicotinic Acetylcholine receptors) in lung cancer (both Non-Small Cell Lung Cancer and Small Cell Lung Cancer) and recapitulate the up-to-date knowledge about their role and clinical relevance for a potential use in the clinical setting, for lung cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Chiara Capitani
- General Pathology Laboratory, Department of Experimental and Clinical Medicine, Internal Medicine Section, University of Florence, Florence, Italy
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Ginevra Chioccioli Altadonna
- General Pathology Laboratory, Department of Experimental and Clinical Medicine, Internal Medicine Section, University of Florence, Florence, Italy
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Michele Santillo
- General Pathology Laboratory, Department of Experimental and Clinical Medicine, Internal Medicine Section, University of Florence, Florence, Italy
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Elena Lastraioli
- General Pathology Laboratory, Department of Experimental and Clinical Medicine, Internal Medicine Section, University of Florence, Florence, Italy
| |
Collapse
|
5
|
Montalbano A, Sala C, Altadonna GC, Becchetti A, Arcangeli A. High throughput clone screening on overexpressed hERG1 and Kv1.3 potassium channels using ion channel reader (ICR) label free technology. Heliyon 2023; 9:e20112. [PMID: 37767500 PMCID: PMC10520782 DOI: 10.1016/j.heliyon.2023.e20112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/27/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Pharmacological studies aimed at the development of newly synthesized drugs directed against ion channels (as well as genetic studies of ion channel mutations) involve the development and use of transfected cells. However, the identification of the best clone, in terms of transfection efficiency, is often a time consuming procedure when performed through traditional methods such as manual patch-clamp. On the other hand, the use of other faster techniques, such as for example the IF, are not informative on the effective biological functionality of the transfected ion channel(s). In the present work, we used the high throughput automated ion channel reader (ICR) technology (ICR8000 Aurora Biomed Inc.) that combine atomic absorption spectroscopy with a patented microsampling process to accurately measure ion flux in cell-based screening assays. This technology indeed helped us to evaluate the transfection efficiency of hERG1 and hKv1.3 channels respectively on the HEK-293 and CHO cellular models. Moreover, as proof of the validity of this innovative method, we have corroborated these data with the functional characterization of the potassium currents carried out by the same clones through patch-clamp recordings. The results obtained in our study are promising and represent a valid methodological strategy to screen a large number of clones simultaneously and to pharmacologically evaluate their functionality within an extremely faster timeframe.
Collapse
Affiliation(s)
- Alberto Montalbano
- Department of Experimental and Clinical Medicine, University of Florence, I-50134, Florence, Italy
| | - Cesare Sala
- Department of Experimental and Clinical Medicine, University of Florence, I-50134, Florence, Italy
| | | | - Andrea Becchetti
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza 2, I-20126, Milano, Italy
| | - Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, University of Florence, I-50134, Florence, Italy
| |
Collapse
|
6
|
Sesti F, Bortolami A, Kathera-Ibarra EF. Non-conducting functions of potassium channels in cancer and neurological disease. CURRENT TOPICS IN MEMBRANES 2023; 92:199-231. [PMID: 38007268 DOI: 10.1016/bs.ctm.2023.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
Cancer and neurodegenerative disease, albeit fundamental differences, share some common pathogenic mechanisms. Accordingly, both conditions are associated with aberrant cell proliferation and migration. Here, we review the causative role played by potassium (K+) channels, a fundamental class of proteins, in cancer and neurodegenerative disease. The concept that emerges from the review of the literature is that K+ channels can promote the development and progression of cancerous and neurodegenerative pathologies by dysregulating cell proliferation and migration. K+ channels appear to control these cellular functions in ways that not necessarily depend on their conducting properties and that involve the ability to directly or indirectly engage growth and survival signaling pathways. As cancer and neurodegenerative disease represent global health concerns, identifying commonalities may help understand the molecular basis for those devastating conditions and may facilitate the design of new drugs or the repurposing of existing drugs.
Collapse
Affiliation(s)
- Federico Sesti
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Hoes Ln. West, Piscataway, NJ, United States.
| | - Alessandro Bortolami
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Hoes Ln. West, Piscataway, NJ, United States
| | - Elena Forzisi Kathera-Ibarra
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Hoes Ln. West, Piscataway, NJ, United States
| |
Collapse
|
7
|
Santini L, Duranti C, Palandri C, Giammarino L, Musumeci M, Carlucci L, Capitani C, Colasurdo R, Recchia F, Cerbai E, Coppini R, Arcangeli A. Cardiac safety assessment of a novel recombinant bispecific antibody targeting the ether-à-go-go related gene 1 (hERG1)-β1 integrin macromolecular complex. Front Pharmacol 2023; 14:1237431. [PMID: 37767396 PMCID: PMC10520717 DOI: 10.3389/fphar.2023.1237431] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/17/2023] [Indexed: 09/29/2023] Open
Abstract
Introduction: In the last decades, mounting evidence has pointed out the human ether-á-go-go-related gene (hERG1) potassium channel as a novel biomarker in human cancers. However, hERG1 sustains the cardiac repolarizing current IKr and its blockade can induce a prolonged QT interval at the ECG, which increases the risk of life-threatening arrhythmias. This represents a major hindrance for targeting hERG1 for antineoplastic therapeutic purposes. Based on our discovery that hERG1 resides in a macromolecular complex with the β1 subunit of integrin adhesion receptors only in tumors, and not in the heart, we generated (and patented WO2019/015936) a novel engineered, single chain, bispecific antibody in the format of a diabody (scDb-hERG1-β1). This antibody has been proven to target with high affinity the hERG1/β1 integrin complex and to exert a good antineoplastic activity in preclinical mouse models. Methods: In the present study, we evaluated the cardiac safety of the scDb-hERG1-β1, determining the action potential duration (APD) of human cardiomyocytes, either atrial (from valve-disease patients) or ventricular (from aortic stenosis patients). Cardiac cells were incubated in vitro with i) the scDb-hERG1-β1, ii) the full length anti-hERG1 monoclonal antibody (mAb-hERG1) and iii) its single chain Fragment variable derivative (scFv-hERG1), from which the scDb-hERG1-β1 was assembled. All the tests were performed before and after treatment with the specific hERG1 blocker E4031. In addition, we have performed preliminary experiments, analyzing the effects of the scDb-hERG1/β1 in vivo measuring the QT interval length of the surface ECG after its injection intravenously in farm-pigs. Results: The scDb-hERG1-β1 did not produce any lengthening of APD compared to control (vehicle) conditions, either in atrial or ventricular cardiomyocytes, whereas both the hERG1-mAb and the scFv-hERG1 produced a significant APD prolongation. The addition of E4031 further prolonged APD. The scDb-hERG1-β1 did not produce any alterations of the QT (and QTc) interval values, once injected intravenously in farm pigs. Discussion: Overall, the above evidences plead for the cardiac safety of the scDb-hERG1-β1, suggesting that an application of this antibody for anti-cancer therapy will be untainted by cardiotoxicity.
Collapse
Affiliation(s)
- Lorenzo Santini
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Claudia Duranti
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence, Florence, Italy
| | - Chiara Palandri
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Lucrezia Giammarino
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Monica Musumeci
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Lucia Carlucci
- Institute of Life Sciences, Scuola Superiore Sant’Anna, Pisa, Italy
| | - Chiara Capitani
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence, Florence, Italy
- Department of Medical Biotechnology, University of Siena, Siena, Italy
| | - Rossella Colasurdo
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence, Florence, Italy
- Department of Medical Biotechnology, University of Siena, Siena, Italy
| | - Fabio Recchia
- Institute of Life Sciences, Scuola Superiore Sant’Anna, Pisa, Italy
| | - Elisabetta Cerbai
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Raffaele Coppini
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence, Florence, Italy
| |
Collapse
|
8
|
Sala C, Staderini M, Lottini T, Duranti C, Angelini G, Constantin G, Arcangeli A. Expression of the ether-a-gò-gò-related gene 1 channel during B and T lymphocyte development: role in BCR and TCR signaling. Front Immunol 2023; 14:1111471. [PMID: 37744334 PMCID: PMC10515723 DOI: 10.3389/fimmu.2023.1111471] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 08/07/2023] [Indexed: 09/26/2023] Open
Abstract
The functional relevance of K+ and Ca2+ ion channels in the "Store Operated Calcium Entry" (SOCE) during B and T lymphocyte activation is well proven. However, their role in the process of T- and B- cell development and selection is still poorly defined. In this scenario, our aim was to characterize the expression of the ether à-go-go-related gene 1 (ERG1) and KV1.3 K+ channels during the early stages of mouse lymphopoiesis and analyze how they affect Ca2+signaling, or other signaling pathways, known to mediate selection and differentiation processes of lymphoid clones. We provide here evidence that the mouse (m)ERG1 is expressed in primary lymphoid organs, bone marrow (BM), and thymus of C57BL/6 and SV129 mice. This expression is particularly evident in the BM during the developmental stages of B cells, before the positive selection (large and small PreB). mERG1 is also expressed in all thymic subsets of both strains, when lymphocyte positive and negative selection occurs. Partially overlapping results were obtained for KV1.3 expression. mERG1 and KV1.3 were expressed at significantly higher levels in B-cell precursors of mice developing an experimental autoimmune encephalomyelitis (EAE). The pharmacological blockage of ERG1 channels with E4031 produced a significant reduction in intracellular Ca2+ after lymphocyte stimulation in the CD4+ and double-positive T-cell precursors' subsets. This suggests that ERG1 might contribute to maintaining the electrochemical gradient responsible for driving Ca2+ entry, during T-cell receptor signaling which sustains lymphocyte selection checkpoints. Such role mirrors that performed by the shaker-type KV1.3 potassium channel during the activation process of mature lymphocytes. No effects on Ca2+ signaling were observed either in B-cell precursors after blocking KV1.3 with PSORA-4. In the BM, the pharmacological blockage of ERG1 channels produced an increase in ERK phosphorylation, suggesting an effect of ERG1 in regulating B-lymphocyte precursor clones' proliferation and checkpoint escape. Overall, our results suggest a novel physiological function of ERG1 in the processes of differentiation and selection of lymphoid precursors, paving the way to further studies aimed at defining the expression and role of ERG1 channels in immune-based pathologies in addition to that during lymphocyte neoplastic transformation.
Collapse
Affiliation(s)
- Cesare Sala
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence, Florence, Italy
| | - Martina Staderini
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence, Florence, Italy
| | - Tiziano Lottini
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence, Florence, Italy
| | - Claudia Duranti
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence, Florence, Italy
| | - Gabriele Angelini
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Gabriela Constantin
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence, Florence, Italy
| |
Collapse
|
9
|
Qin W, Li YH, Tong J, Wu J, Zhao D, Li HJ, Xing L, He CX, Zhou X, Li PQ, Meng G, Wu SP, Cao HL. Rational Design and Synthesis of 3-Morpholine Linked Aromatic-Imino-1H-Indoles as Novel Kv1.5 Channel Inhibitors Sharing Vasodilation Effects. Front Mol Biosci 2022; 8:805594. [PMID: 35141279 PMCID: PMC8819089 DOI: 10.3389/fmolb.2021.805594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 12/28/2021] [Indexed: 11/25/2022] Open
Abstract
Atrial fibrillation (AF) is the most common clinical sustained arrhythmia; clinical therapeutic drugs have low atrial selectivity and might cause more severe ventricle arrhythmias while stopping AF. As an anti-AF drug target with high selectivity on the atrial muscle cells, the undetermined crystal structure of Kv1.5 potassium channel impeded further new drug development. Herein, with the simulated 3D structure of Kv1.5 as the drug target, a series of 3-morpholine linked aromatic amino substituted 1H-indoles as novel Kv1.5 channel inhibitors were designed and synthesized based on target–ligand interaction analysis. The synthesis route was practical, starting from commercially available material, and the chemical structures of target compounds were characterized. It was indicated that compounds T16 and T5 (100 μM) exhibited favorable inhibitory activity against the Kv1.5 channel with an inhibition rate of 70.8 and 57.5% using a patch clamp technique. All compounds did not exhibit off-target effects against other drug targets, which denoted some selectivity on the Kv1.5 channel. Interestingly, twelve compounds exhibited favorable vasodilation activity on pre-contracted arterial rings in vitro using KCl or phenylephrine (PE) by a Myograph. The vasodilation rates of compounds T16 and T4 (100 μM) even reached over 90%, which would provide potential lead compounds for both anti-AF and anti-hypertension new drug development.
Collapse
Affiliation(s)
- Wei Qin
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Yi-Heng Li
- College of Life Sciences, Northwest University, Xi’an, China
| | - Jing Tong
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Jie Wu
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Dong Zhao
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Hui-Jin Li
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Lu Xing
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Chun-Xia He
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Xin Zhou
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Peng-Quan Li
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Ge Meng
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, China
- Engineering Center of Catalysis and Synthesis for Chiral Molecules, Shanghai Engineering Center of Industrial Asymmetric Catalysis for Chiral Drugs, Fudan University, Shanghai, China
- *Correspondence: Ge Meng, ; Shao-Ping Wu, ; Hui-Ling Cao,
| | - Shao-Ping Wu
- College of Life Sciences, Northwest University, Xi’an, China
- *Correspondence: Ge Meng, ; Shao-Ping Wu, ; Hui-Ling Cao,
| | - Hui-Ling Cao
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
- College of Life Sciences, Northwest University, Xi’an, China
- *Correspondence: Ge Meng, ; Shao-Ping Wu, ; Hui-Ling Cao,
| |
Collapse
|
10
|
Allred TK, Shaghafi MB, Chen PP, Tran Q, Houk KN, Overman LE. Constructing Saturated Guanidinum Heterocycles by Cycloaddition of N-Amidinyliminium Ions with Indoles. Org Lett 2021; 23:7618-7623. [PMID: 34546759 DOI: 10.1021/acs.orglett.1c02832] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We report that structurally complex guanidinium heterocycles can be prepared in one step by regio- and stereoselective [4 + 2]-cycloadditions of N-amidinyliminium ions with indoles or benzothiophene. In contrast to reactions of these heterodienes with alkenes, density functional theory (DFT) calculations show that these cycloadditions take place in a concerted asynchronous fashion. The [4 + 2]-cycloaddition of N-amidinyliminium ions (1,3-diaza-1,3-dienes) with indoles and benzothiophene are distinctive, as related [4 + 2]-cycloadditions of N-acyliminium ions (1-oxa-3-aza-1,3-dienes) are apparently unknown.
Collapse
Affiliation(s)
- Tyler K Allred
- Department of Chemistry, University of California, Irvine, California 92697-2025, United States
| | - Michael B Shaghafi
- Department of Chemistry, University of California, Irvine, California 92697-2025, United States
| | - Pan-Pan Chen
- Department of Chemistry & Biochemistry, University of California, Los Angeles, California 90095-1569, United States
| | - Quan Tran
- Department of Chemistry & Biochemistry, University of California, Los Angeles, California 90095-1569, United States
| | - K N Houk
- Department of Chemistry & Biochemistry, University of California, Los Angeles, California 90095-1569, United States
| | - Larry E Overman
- Department of Chemistry, University of California, Irvine, California 92697-2025, United States
| |
Collapse
|
11
|
Cirri D, Bartoli F, Pratesi A, Baglini E, Barresi E, Marzo T. Strategies for the Improvement of Metal-Based Chemotherapeutic Treatments. Biomedicines 2021; 9:504. [PMID: 34064364 PMCID: PMC8147839 DOI: 10.3390/biomedicines9050504] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 12/12/2022] Open
Abstract
This article provides an overview of the various research approaches we have explored in recent years to improve metal-based agents for cancer or infection treatments. Although cisplatin, carboplatin, and oxaliplatin remain the cornerstones in tumor chemotherapy, the discovery and approval of novel inorganic anticancer drugs is a very slow process. Analogously, although a few promising inorganic drugs have found clinical application against parasitic or bacterial infections, their use remains relatively limited. Moreover, the discovery process is often affected by small therapeutic enhancements that are not attractive for the pharmaceutical industry. However, the availability of increasing mechanistic information for the modes of action of established inorganic drugs is fueling the exploration of various approaches for developing effective inorganic chemotherapy agents. Through a series of examples, some from our own research experience, we focus our attention on a number of promising strategies, including (1) drug repurposing, (2) the simple modification of the chemical structures of approved metal-based drugs, (3) testing novel drug combinations, and (4) newly synthesized complexes coupling different anticancer drugs. Accordingly, we aim to suggest and summarize a series of reliable approaches that are exploitable for the development of improved and innovative treatments.
Collapse
Affiliation(s)
- Damiano Cirri
- Department of Chemistry and Industrial Chemistry (DCCI), Univerisity of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy;
| | - Francesco Bartoli
- Department of Translational Research and of New Surgical and Medical Technologies, Univerisity of Pisa, Via Risorgimento, 36, 56126 Pisa, Italy;
| | - Alessandro Pratesi
- Department of Chemistry and Industrial Chemistry (DCCI), Univerisity of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy;
| | - Emma Baglini
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy; (E.B.); (E.B.)
| | - Elisabetta Barresi
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy; (E.B.); (E.B.)
| | - Tiziano Marzo
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy; (E.B.); (E.B.)
| |
Collapse
|
12
|
Bachmann M, Li W, Edwards MJ, Ahmad SA, Patel S, Szabo I, Gulbins E. Voltage-Gated Potassium Channels as Regulators of Cell Death. Front Cell Dev Biol 2020; 8:611853. [PMID: 33381507 PMCID: PMC7767978 DOI: 10.3389/fcell.2020.611853] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
Ion channels allow the flux of specific ions across biological membranes, thereby determining ion homeostasis within the cells. Voltage-gated potassium-selective ion channels crucially contribute to the setting of the plasma membrane potential, to volume regulation and to the physiologically relevant modulation of intracellular potassium concentration. In turn, these factors affect cell cycle progression, proliferation and apoptosis. The present review summarizes our current knowledge about the involvement of various voltage-gated channels of the Kv family in the above processes and discusses the possibility of their pharmacological targeting in the context of cancer with special emphasis on Kv1.1, Kv1.3, Kv1.5, Kv2.1, Kv10.1, and Kv11.1.
Collapse
Affiliation(s)
- Magdalena Bachmann
- Department of Biology, University of Padova, Padua, Italy.,Department of Surgery, Medical School, University of Cincinnati, Cincinnati, OH, United States
| | - Weiwei Li
- Department of Surgery, Medical School, University of Cincinnati, Cincinnati, OH, United States
| | - Michael J Edwards
- Department of Surgery, Medical School, University of Cincinnati, Cincinnati, OH, United States
| | - Syed A Ahmad
- Department of Surgery, Medical School, University of Cincinnati, Cincinnati, OH, United States
| | - Sameer Patel
- Department of Surgery, Medical School, University of Cincinnati, Cincinnati, OH, United States
| | - Ildiko Szabo
- Department of Biology, University of Padova, Padua, Italy.,Consiglio Nazionale delle Ricerche Institute of Neuroscience, Padua, Italy
| | - Erich Gulbins
- Department of Surgery, Medical School, University of Cincinnati, Cincinnati, OH, United States.,Department of Molecular Biology, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
13
|
Novel Therapeutic Approaches of Ion Channels and Transporters in Cancer. Rev Physiol Biochem Pharmacol 2020; 183:45-101. [PMID: 32715321 DOI: 10.1007/112_2020_28] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The expression and function of many ion channels and transporters in cancer cells display major differences in comparison to those from healthy cells. These differences provide the cancer cells with advantages for tumor development. Accordingly, targeting ion channels and transporters have beneficial anticancer effects including inhibition of cancer cell proliferation, migration, invasion, metastasis, tumor vascularization, and chemotherapy resistance, as well as promoting apoptosis. Some of the molecular mechanisms associating ion channels and transporters with cancer include the participation of oxidative stress, immune response, metabolic pathways, drug synergism, as well as noncanonical functions of ion channels. This diversity of mechanisms offers an exciting possibility to suggest novel and more effective therapeutic approaches to fight cancer. Here, we review and discuss most of the current knowledge suggesting novel therapeutic approaches for cancer therapy targeting ion channels and transporters. The role and regulation of ion channels and transporters in cancer provide a plethora of exceptional opportunities in drug design, as well as novel and promising therapeutic approaches that may be used for the benefit of cancer patients.
Collapse
|
14
|
Palme D, Misovic M, Ganser K, Klumpp L, Salih HR, Zips D, Huber SM. hERG K + Channels Promote Survival of Irradiated Leukemia Cells. Front Pharmacol 2020; 11:489. [PMID: 32390841 PMCID: PMC7194033 DOI: 10.3389/fphar.2020.00489] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/27/2020] [Indexed: 12/19/2022] Open
Abstract
Many tumor cells express highly elevated activities of voltage-gated K+ channels in the plasma membrane which are indispensable for tumor growth. To test for K+ channel function during DNA damage response, we subjected human chronic myeloid leukemia (CML) cells to sub-lethal doses of ionizing radiation (0-8 Gy, 6 MV photons) and determined K+ channel activity, K+ channel-dependent Ca2+ signaling, cell cycle progression, DNA repair, and clonogenic survival by whole-cell patch clamp recording, fura-2 Ca2+ imaging, Western blotting, flow cytometry, immunofluorescence microscopy, and pre-plating colony formation assay, respectively. As a result, the human erythroid CML cell line K562 and primary human CML cells functionally expressed hERG1. Irradiation stimulated in both cell types an increase in the activity of hERG1 K+ channels which became apparent 1-2 h post-irradiation. This increase in K+ channel activity was paralleled by an accumulation in S phase of cell cycle followed by a G2/M cell cycle arrest as analyzed between 8 and 72 h post-irradiation. Attenuating the K+ channel function by applying the hERG1 channel inhibitor E4031 modulated Ca2+ signaling, impaired inhibition of the mitosis promoting subunit cdc2, overrode cell cycle arrest, and decreased clonogenic survival of the irradiated cells but did not affect repair of DNA double strand breaks suggesting a critical role of the hERG1 K+ channels for the Ca2+ signaling and the cell cycle control during DNA damage response.
Collapse
Affiliation(s)
- Daniela Palme
- Department of Radiation Oncology, University Hospital Tübingen, Tübingen, Germany
| | - Milan Misovic
- Department of Radiation Oncology, University Hospital Tübingen, Tübingen, Germany
| | - Katrin Ganser
- Department of Radiation Oncology, University Hospital Tübingen, Tübingen, Germany
| | - Lukas Klumpp
- Department of Radiation Oncology, University Hospital Tübingen, Tübingen, Germany
| | - Helmut R Salih
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), University Hospital Tübingen, Tübingen, Germany
| | - Daniel Zips
- Department of Radiation Oncology, University Hospital Tübingen, Tübingen, Germany.,German Cancer Consortium (DKTK), Partner Site Tübingen, Tübingen, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephan M Huber
- Department of Radiation Oncology, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
15
|
Zhao Z, Ruan S, Ma X, Feng Q, Xie Z, Nie Z, Fan P, Qian M, He X, Wu S, Zhang Y, Zheng X. Challenges Faced with Small Molecular Modulators of Potassium Current Channel Isoform Kv1.5. Biomolecules 2019; 10:E10. [PMID: 31861703 PMCID: PMC7022446 DOI: 10.3390/biom10010010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/08/2019] [Accepted: 12/10/2019] [Indexed: 12/30/2022] Open
Abstract
The voltage-gated potassium channel Kv1.5, which mediates the cardiac ultra-rapid delayed-rectifier (IKur) current in human cells, has a crucial role in atrial fibrillation. Therefore, the design of selective Kv1.5 modulators is essential for the treatment of pathophysiological conditions involving Kv1.5 activity. This review summarizes the progress of molecular structures and the functionality of different types of Kv1.5 modulators, with a focus on clinical cardiovascular drugs and a number of active natural products, through a summarization of 96 compounds currently widely used. Furthermore, we also discuss the contributions of Kv1.5 and the regulation of the structure-activity relationship (SAR) of synthetic Kv1.5 inhibitors in human pathophysiology. SAR analysis is regarded as a useful strategy in structural elucidation, as it relates to the characteristics that improve compounds targeting Kv1.5. Herein, we present previous studies regarding the structural, pharmacological, and SAR information of the Kv1.5 modulator, through which we can assist in identifying and designing potent and specific Kv1.5 inhibitors in the treatment of diseases involving Kv1.5 activity.
Collapse
Affiliation(s)
- Zefeng Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, 229 Taibai Road, Xi’an 710069, China; (Z.Z.); (S.R.); (X.M.); (Q.F.); (Z.X.); (Z.N.); (P.F.); (Y.Z.); (X.Z.)
- Biomedicine Key Laboratory of Shaanxi Province, School of Pharmacy, Northwest University, 229 Taibai Road, Xi’an 710069, China
| | - Songsong Ruan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, 229 Taibai Road, Xi’an 710069, China; (Z.Z.); (S.R.); (X.M.); (Q.F.); (Z.X.); (Z.N.); (P.F.); (Y.Z.); (X.Z.)
- Biomedicine Key Laboratory of Shaanxi Province, School of Pharmacy, Northwest University, 229 Taibai Road, Xi’an 710069, China
| | - Xiaoming Ma
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, 229 Taibai Road, Xi’an 710069, China; (Z.Z.); (S.R.); (X.M.); (Q.F.); (Z.X.); (Z.N.); (P.F.); (Y.Z.); (X.Z.)
- Biomedicine Key Laboratory of Shaanxi Province, School of Pharmacy, Northwest University, 229 Taibai Road, Xi’an 710069, China
| | - Qian Feng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, 229 Taibai Road, Xi’an 710069, China; (Z.Z.); (S.R.); (X.M.); (Q.F.); (Z.X.); (Z.N.); (P.F.); (Y.Z.); (X.Z.)
- Biomedicine Key Laboratory of Shaanxi Province, School of Pharmacy, Northwest University, 229 Taibai Road, Xi’an 710069, China
| | - Zhuosong Xie
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, 229 Taibai Road, Xi’an 710069, China; (Z.Z.); (S.R.); (X.M.); (Q.F.); (Z.X.); (Z.N.); (P.F.); (Y.Z.); (X.Z.)
- Biomedicine Key Laboratory of Shaanxi Province, School of Pharmacy, Northwest University, 229 Taibai Road, Xi’an 710069, China
| | - Zhuang Nie
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, 229 Taibai Road, Xi’an 710069, China; (Z.Z.); (S.R.); (X.M.); (Q.F.); (Z.X.); (Z.N.); (P.F.); (Y.Z.); (X.Z.)
- Biomedicine Key Laboratory of Shaanxi Province, School of Pharmacy, Northwest University, 229 Taibai Road, Xi’an 710069, China
| | - Peinan Fan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, 229 Taibai Road, Xi’an 710069, China; (Z.Z.); (S.R.); (X.M.); (Q.F.); (Z.X.); (Z.N.); (P.F.); (Y.Z.); (X.Z.)
- Biomedicine Key Laboratory of Shaanxi Province, School of Pharmacy, Northwest University, 229 Taibai Road, Xi’an 710069, China
| | - Mingcheng Qian
- Department of Medicinal Chemistry, School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou 213164, China;
- Laboratory for Medicinal Chemistry, Ghent University, Ottergemsesteenweg 460, B-9000 Ghent, Belgium
| | - Xirui He
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, China;
| | - Shaoping Wu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, 229 Taibai Road, Xi’an 710069, China; (Z.Z.); (S.R.); (X.M.); (Q.F.); (Z.X.); (Z.N.); (P.F.); (Y.Z.); (X.Z.)
- Biomedicine Key Laboratory of Shaanxi Province, School of Pharmacy, Northwest University, 229 Taibai Road, Xi’an 710069, China
| | - Yongmin Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, 229 Taibai Road, Xi’an 710069, China; (Z.Z.); (S.R.); (X.M.); (Q.F.); (Z.X.); (Z.N.); (P.F.); (Y.Z.); (X.Z.)
- Biomedicine Key Laboratory of Shaanxi Province, School of Pharmacy, Northwest University, 229 Taibai Road, Xi’an 710069, China
- Sorbonne Université, Institut Parisien de Chimie Moléculaire, CNRS UMR 8232, 4 place Jussieu, 75005 Paris, France
| | - Xiaohui Zheng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, 229 Taibai Road, Xi’an 710069, China; (Z.Z.); (S.R.); (X.M.); (Q.F.); (Z.X.); (Z.N.); (P.F.); (Y.Z.); (X.Z.)
- Biomedicine Key Laboratory of Shaanxi Province, School of Pharmacy, Northwest University, 229 Taibai Road, Xi’an 710069, China
| |
Collapse
|
16
|
Hutchings CJ, Colussi P, Clark TG. Ion channels as therapeutic antibody targets. MAbs 2018; 11:265-296. [PMID: 30526315 PMCID: PMC6380435 DOI: 10.1080/19420862.2018.1548232] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 11/01/2018] [Accepted: 11/03/2018] [Indexed: 12/12/2022] Open
Abstract
It is now well established that antibodies have numerous potential benefits when developed as therapeutics. Here, we evaluate the technical challenges of raising antibodies to membrane-spanning proteins together with enabling technologies that may facilitate the discovery of antibody therapeutics to ion channels. Additionally, we discuss the potential targeting opportunities in the anti-ion channel antibody landscape, along with a number of case studies where functional antibodies that target ion channels have been reported. Antibodies currently in development and progressing towards the clinic are highlighted.
Collapse
Affiliation(s)
| | | | - Theodore G. Clark
- TetraGenetics Inc, Arlington Massachusetts, USA
- Department of Microbiology and Immunology, Cornell University, Ithaca New York, USA
| |
Collapse
|
17
|
The combined activation of K Ca3.1 and inhibition of K v11.1/hERG1 currents contribute to overcome Cisplatin resistance in colorectal cancer cells. Br J Cancer 2017; 118:200-212. [PMID: 29161243 PMCID: PMC5785745 DOI: 10.1038/bjc.2017.392] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Revised: 09/25/2017] [Accepted: 10/02/2017] [Indexed: 02/06/2023] Open
Abstract
Background: Platinum-based drugs such as Cisplatin are commonly employed for cancer treatment. Despite an initial therapeutic response, Cisplatin treatment often results in the development of chemoresistance. To identify novel approaches to overcome Cisplatin resistance, we tested Cisplatin in combination with K+ channel modulators on colorectal cancer (CRC) cells. Methods: The functional expression of Ca2+-activated (KCa3.1, also known as KCNN4) and voltage-dependent (Kv11.1, also known as KCNH2 or hERG1) K+ channels was determined in two CRC cell lines (HCT-116 and HCT-8) by molecular and electrophysiological techniques. Cisplatin and several K+ channel modulators were tested in vitro for their action on K+ currents, cell vitality, apoptosis, cell cycle, proliferation, intracellular signalling and Platinum uptake. These effects were also analysed in a mouse model mimicking Cisplatin resistance. Results: Cisplatin-resistant CRC cells expressed higher levels of KCa3.1 and Kv11.1 channels, compared with Cisplatin-sensitive CRC cells. In resistant cells, KCa3.1 activators (SKA-31) and Kv11.1 inhibitors (E4031) had a synergistic action with Cisplatin in triggering apoptosis and inhibiting proliferation. The effect was maximal when KCa3.1 activation and Kv11.1 inhibition were combined. In fact, similar results were produced by Riluzole, which is able to both activate KCa3.1 and inhibit Kv11.1. Cisplatin uptake into resistant cells depended on KCa3.1 channel activity, as it was potentiated by KCa3.1 activators. Kv11.1 blockade led to increased KCa3.1 expression and thereby stimulated Cisplatin uptake. Finally, the combined administration of a KCa3.1 activator and a Kv11.1 inhibitor also overcame Cisplatin resistance in vivo. Conclusions: As Riluzole, an activator of KCa3.1 and inhibitor of Kv11.1 channels, is in clinical use, our results suggest that this compound may be useful in the clinic to improve Cisplatin efficacy and overcome Cisplatin resistance in CRC.
Collapse
|
18
|
Becchetti A, Crescioli S, Zanieri F, Petroni G, Mercatelli R, Coppola S, Gasparoli L, D'Amico M, Pillozzi S, Crociani O, Stefanini M, Fiore A, Carraresi L, Morello V, Manoli S, Brizzi MF, Ricci D, Rinaldi M, Masi A, Schmidt T, Quercioli F, Defilippi P, Arcangeli A. The conformational state of hERG1 channels determines integrin association, downstream signaling, and cancer progression. Sci Signal 2017; 10:10/473/eaaf3236. [PMID: 28377405 DOI: 10.1126/scisignal.aaf3236] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Ion channels regulate cell proliferation, differentiation, and migration in normal and neoplastic cells through cell-cell and cell-extracellular matrix (ECM) transmembrane receptors called integrins. K+ flux through the human ether-à-go-go-related gene 1 (hERG1) channel shapes action potential firing in excitable cells such as cardiomyocytes. Its abundance is often aberrantly high in tumors, where it modulates integrin-mediated signaling. We found that hERG1 interacted with the β1 integrin subunit at the plasma membrane of human cancer cells. This interaction was not detected in cardiomyocytes because of the presence of the hERG1 auxiliary subunit KCNE1 (potassium voltage-gated channel subfamily E regulatory subunit 1), which blocked the β1 integrin-hERG1 interaction. Although open hERG1 channels did not interact as strongly with β1 integrins as did closed channels, current flow through hERG1 channels was necessary to activate the integrin-dependent phosphorylation of Tyr397 in focal adhesion kinase (FAK) in both normal and cancer cells. In immunodeficient mice, proliferation was inhibited in breast cancer cells expressing forms of hERG1 with impaired K+ flow, whereas metastasis of breast cancer cells was reduced when the hERG1/β1 integrin interaction was disrupted. We conclude that the interaction of β1 integrins with hERG1 channels in cancer cells stimulated distinct signaling pathways that depended on the conformational state of hERG1 and affected different aspects of tumor progression.
Collapse
Affiliation(s)
- Andrea Becchetti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milano, Italy
| | - Silvia Crescioli
- Department of Experimental and Clinical Medicine, University of Firenze, Viale G.B. Morgagni 50, 50134 Firenze, Italy
| | - Francesca Zanieri
- Department of Experimental and Clinical Medicine, University of Firenze, Viale G.B. Morgagni 50, 50134 Firenze, Italy
| | - Giulia Petroni
- Department of Experimental and Clinical Medicine, University of Firenze, Viale G.B. Morgagni 50, 50134 Firenze, Italy
| | - Raffaella Mercatelli
- Consiglio Nazionale delle Ricerche-Istituto Nazionale di Ottica, Via N. Carrara 1, 50019 Sesto Fiorentino, Italy
| | - Stefano Coppola
- Physics of Life Processes, Huygens-Kamerlingh Onnes Laboratory, Leiden University, Niels Bohrweg 2, 2333 CA Leiden, Netherlands
| | - Luca Gasparoli
- Department of Experimental and Clinical Medicine, University of Firenze, Viale G.B. Morgagni 50, 50134 Firenze, Italy
| | - Massimo D'Amico
- Di.V.A.L. Toscana SRL, Via Madonna del Piano 6, 50019 Sesto Fiorentino, Italy
| | - Serena Pillozzi
- Department of Experimental and Clinical Medicine, University of Firenze, Viale G.B. Morgagni 50, 50134 Firenze, Italy
| | - Olivia Crociani
- Department of Experimental and Clinical Medicine, University of Firenze, Viale G.B. Morgagni 50, 50134 Firenze, Italy
| | - Matteo Stefanini
- Di.V.A.L. Toscana SRL, Via Madonna del Piano 6, 50019 Sesto Fiorentino, Italy
| | - Antonella Fiore
- Department of Experimental and Clinical Medicine, University of Firenze, Viale G.B. Morgagni 50, 50134 Firenze, Italy
| | - Laura Carraresi
- Di.V.A.L. Toscana SRL, Via Madonna del Piano 6, 50019 Sesto Fiorentino, Italy
| | - Virginia Morello
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - Sagar Manoli
- Department of Experimental and Clinical Medicine, University of Firenze, Viale G.B. Morgagni 50, 50134 Firenze, Italy
| | - Maria Felice Brizzi
- Department of Medical Sciences, University of Torino, Corso Dogliotti 14, 10126 Torino, Italy
| | - Davide Ricci
- Department of Surgical Sciences, University of Torino, Corso Dogliotti 14, 10126 Torino, Italy
| | - Mauro Rinaldi
- Department of Surgical Sciences, University of Torino, Corso Dogliotti 14, 10126 Torino, Italy
| | - Alessio Masi
- Department of Experimental and Clinical Medicine, University of Firenze, Viale G.B. Morgagni 50, 50134 Firenze, Italy
| | - Thomas Schmidt
- Physics of Life Processes, Huygens-Kamerlingh Onnes Laboratory, Leiden University, Niels Bohrweg 2, 2333 CA Leiden, Netherlands
| | - Franco Quercioli
- Consiglio Nazionale delle Ricerche-Istituto Nazionale di Ottica, Via N. Carrara 1, 50019 Sesto Fiorentino, Italy
| | - Paola Defilippi
- Physics of Life Processes, Huygens-Kamerlingh Onnes Laboratory, Leiden University, Niels Bohrweg 2, 2333 CA Leiden, Netherlands
| | - Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, University of Firenze, Viale G.B. Morgagni 50, 50134 Firenze, Italy.
| |
Collapse
|
19
|
Arcangeli A, Becchetti A. hERG Channels: From Antitargets to Novel Targets for Cancer Therapy. Clin Cancer Res 2016; 23:3-5. [PMID: 27903676 DOI: 10.1158/1078-0432.ccr-16-2322] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 10/21/2016] [Accepted: 10/27/2016] [Indexed: 01/12/2023]
Abstract
In this issue of Clinical Cancer Research, evidence is provided on how to avoid cardiotoxicity when targeting hERG K+ channel for cancer therapy. hERG regulates different aspects of neoplastic progression. Although its blockade has effective anticancer effects in experimental models, it may lead to fatal arrhythmias in humans. Clin Cancer Res; 23(1); 3-5. ©2016 AACRSee related article by Pointer et al., p. 73.
Collapse
Affiliation(s)
- Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
| | - Andrea Becchetti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
20
|
Pointer KB, Clark PA, Eliceiri KW, Salamat MS, Robertson GA, Kuo JS. Administration of Non-Torsadogenic human Ether-à-go-go-Related Gene Inhibitors Is Associated with Better Survival for High hERG-Expressing Glioblastoma Patients. Clin Cancer Res 2016; 23:73-80. [PMID: 27635088 DOI: 10.1158/1078-0432.ccr-15-3169] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 08/12/2016] [Accepted: 09/03/2016] [Indexed: 12/29/2022]
Abstract
PURPOSE Glioblastoma is the most malignant primary brain tumor, with a median survival of less than 2 years. More effective therapeutic approaches are needed to improve clinical outcomes. EXPERIMENTAL DESIGN Glioblastoma patient-derived cells (GPDC) were isolated from patient glioblastomas and implanted in mice to form xenografts. IHC was performed for human Ether-à-go-go-Related Gene (hERG) expression and tumor proliferation. Sphere-forming assays with the hERG blocker E-4031 were performed on a high and low hERG-expressing lines. A glioblastoma tissue microarray (TMA; 115 patients) was used to correlate hERG expression with patient survival. Clinical data were analyzed to determine whether patient survival was affected by incidental administration of hERG inhibitory drugs and the correlative effect of patient glioblastoma hERG expression levels. RESULTS hERG expression was upregulated in glioblastoma xenografts with higher proliferative indices. High hERG-expressing GPDCs showed a reduction in sphere formation when treated with hERG inhibitors compared with low hERG-expressing GPDCs. Glioblastoma TMA analysis showed worse survival for glioblastoma patients with high hERG expression versus low expression-43.5 weeks versus 60.9 weeks, respectively (P = 0.022). Furthermore, patients who received at least one hERG blocker had a better survival rate compared with patients who did not (P = 0.0015). Subgroup analysis showed that glioblastoma patients with high hERG expression who received hERG blockers had improved survival (P = 0.0458). There was no difference in survival for low hERG-expressing glioblastoma patients who received hERG blockers (P = 0.4136). CONCLUSIONS Our findings suggest that hERG is a potential glioblastoma survival marker, and that already approved drugs with non-torsadogenic hERG inhibitory activity may potentially be repurposed as adjuvant glioblastoma therapy in high hERG-expressing glioblastoma patients. Clin Cancer Res; 23(1); 73-80. ©2016 AACRSee related commentary by Arcangeli and Becchetti, p. 3.
Collapse
Affiliation(s)
- Kelli B Pointer
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin.,Medical Scientist Training Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin.,Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin.,Laboratory for Optical and Computational Instrumentation (LOCI), University of Wisconsin-Madison, Madison, Wisconsin
| | - Paul A Clark
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kevin W Eliceiri
- Laboratory for Optical and Computational Instrumentation (LOCI), University of Wisconsin-Madison, Madison, Wisconsin.,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - M Shahriar Salamat
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Gail A Robertson
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin. .,Cardiovascular Research Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - John S Kuo
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin. .,Medical Scientist Training Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin.,Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin.,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin.,Department of Surgery, National University of Singapore, Singapore
| |
Collapse
|
21
|
Pharmacological targeting of ion channels for cancer therapy: In vivo evidences. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1385-97. [DOI: 10.1016/j.bbamcr.2015.11.032] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 11/25/2015] [Accepted: 11/26/2015] [Indexed: 12/29/2022]
|
22
|
Macrolide antibiotics exert antileukemic effects by modulating the autophagic flux through inhibition of hERG1 potassium channels. Blood Cancer J 2016; 6:e423. [PMID: 27176799 PMCID: PMC4916301 DOI: 10.1038/bcj.2016.32] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
23
|
Spadavecchia J, Movia D, Moore C, Maguire CM, Moustaoui H, Casale S, Volkov Y, Prina-Mello A. Targeted polyethylene glycol gold nanoparticles for the treatment of pancreatic cancer: from synthesis to proof-of-concept in vitro studies. Int J Nanomedicine 2016; 11:791-822. [PMID: 27013874 PMCID: PMC4777276 DOI: 10.2147/ijn.s97476] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The main objective of this study was to optimize and characterize a drug delivery carrier for doxorubicin, intended to be intravenously administered, capable of improving the therapeutic index of the chemotherapeutic agent itself, and aimed at the treatment of pancreatic cancer. In light of this goal, we report a robust one-step method for the synthesis of dicarboxylic acid-terminated polyethylene glycol (PEG)-gold nanoparticles (AuNPs) and doxorubicin-loaded PEG-AuNPs, and their further antibody targeting (anti-Kv11.1 polyclonal antibody [pAb]). In in vitro proof-of-concept studies, we evaluated the influence of the nanocarrier and of the active targeting functionality on the anti-tumor efficacy of doxorubicin, with respect to its half-maximal effective concentration (EC50) and drug-triggered changes in the cell cycle. Our results demonstrated that the therapeutic efficacy of doxorubicin was positively influenced not only by the active targeting exploited through anti-Kv11.1-pAb but also by the drug coupling with a nanometer-sized delivery system, which indeed resulted in a 30-fold decrease of doxorubicin EC50, cell cycle blockage, and drug localization in the cell nuclei. The cell internalization pathway was strongly influenced by the active targeting of the Kv11.1 subunit of the human Ether-à-go-go related gene 1 (hERG1) channel aberrantly expressed on the membrane of pancreatic cancer cells. Targeted PEG-AuNPs were translocated into the lysosomes and were associated to an increased lysosomal function in PANC-1 cells. Additionally, doxorubicin release into an aqueous environment was almost negligible after 7 days, suggesting that drug release from PEG-AuNPs was triggered by enzymatic activity. Although preliminary, data gathered from this study have considerable potential in the application of safe-by-design nano-enabled drug-delivery systems (ie, nanomedicines) for the treatment of pancreatic cancer, a disease with a poor prognosis and one of the main current burdens of today's health care bill of industrialized countries.
Collapse
Affiliation(s)
- Jolanda Spadavecchia
- Laboratoire de Réactivité de Surface, Sorbonne Universités, UPMC Univ Paris VI, Paris
- Centre National de la recherche française, UMR 7244, CSPBAT, Laboratory of Chemistry, Structures, and Properties of Biomaterials and Therapeutic Agents, Université Paris 13, Sorbonne Paris Cité, Bobigny, France
| | - Dania Movia
- AMBER Centre, CRANN Institute, Dublin, Ireland
| | - Caroline Moore
- AMBER Centre, CRANN Institute, Dublin, Ireland
- Department of Clinical Medicine, School of Medicine, Trinity College, Dublin, Ireland
| | - Ciaran Manus Maguire
- AMBER Centre, CRANN Institute, Dublin, Ireland
- Department of Clinical Medicine, School of Medicine, Trinity College, Dublin, Ireland
| | - Hanane Moustaoui
- Centre National de la recherche française, UMR 7244, CSPBAT, Laboratory of Chemistry, Structures, and Properties of Biomaterials and Therapeutic Agents, Université Paris 13, Sorbonne Paris Cité, Bobigny, France
| | - Sandra Casale
- Laboratoire de Réactivité de Surface, Sorbonne Universités, UPMC Univ Paris VI, Paris
| | - Yuri Volkov
- AMBER Centre, CRANN Institute, Dublin, Ireland
- Department of Clinical Medicine, School of Medicine, Trinity College, Dublin, Ireland
| | - Adriele Prina-Mello
- AMBER Centre, CRANN Institute, Dublin, Ireland
- Department of Clinical Medicine, School of Medicine, Trinity College, Dublin, Ireland
| |
Collapse
|
24
|
Arcangeli A, Becchetti A. Novel perspectives in cancer therapy: Targeting ion channels. Drug Resist Updat 2015; 21-22:11-9. [DOI: 10.1016/j.drup.2015.06.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 06/24/2015] [Accepted: 06/27/2015] [Indexed: 01/04/2023]
|
25
|
Lastraioli E, Perrone G, Sette A, Fiore A, Crociani O, Manoli S, D'Amico M, Masselli M, Iorio J, Callea M, Borzomati D, Nappo G, Bartolozzi F, Santini D, Bencini L, Farsi M, Boni L, Di Costanzo F, Schwab A, Onetti Muda A, Coppola R, Arcangeli A. hERG1 channels drive tumour malignancy and may serve as prognostic factor in pancreatic ductal adenocarcinoma. Br J Cancer 2015; 112:1076-1087. [PMID: 25719829 PMCID: PMC4366888 DOI: 10.1038/bjc.2015.28] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 01/02/2015] [Accepted: 01/12/2015] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND hERG1 channels are aberrantly expressed in human cancers. The expression, functional role and clinical significance of hERG1 channels in pancreatic ductal adenocarcinoma (PDAC) is lacking. METHODS hERG1 expression was tested in PDAC primary samples assembled as tissue microarray by immunohistochemistry using an anti-hERG1 monoclonal antibody (α-hERG1-MoAb). The functional role of hERG1 was studied in PDAC cell lines and primary cultures. ERG1 expression during PDAC progression was studied in Pdx-1-Cre,LSL-Kras(G12D/+),LSL-Trp53(R175H/+) transgenic (KPC) mice. ERG1 expression in vivo was determined by optical imaging using Alexa-680-labelled α-hERG1-MoAb. RESULTS (i) hERG1 was expressed at high levels in 59% of primary PDAC; (ii) hERG1 blockade decreased PDAC cell growth and migration; (iii) hERG1 was physically and functionally linked to the Epidermal Growth Factor-Receptor pathway; (iv) in transgenic mice, ERG1 was expressed in PanIN lesions, reaching high expression levels in PDAC; (v) PDAC patients whose primary tumour showed high hERG1 expression had a worse prognosis; (vi) the α-hERG1-MoAb could detect PDAC in vivo. CONCLUSIONS hERG1 regulates PDAC malignancy and its expression, once validated in a larger cohort also comprising of late-stage, non-surgically resected cases, may be exploited for diagnostic and prognostic purposes in PDAC either ex vivo or in vivo.
Collapse
Affiliation(s)
- E Lastraioli
- Department of Experimental and Clinical Medicine, University of Florence, Viale GB Morgagni 50, Florence 50134, Italy
| | - G Perrone
- Department of Pathology, Pathology Unit, Campus Bio-Medico University, via del Portillo 200, Rome 00128, Italy
| | - A Sette
- Department of Experimental and Clinical Medicine, University of Florence, Viale GB Morgagni 50, Florence 50134, Italy
| | - A Fiore
- Department of Experimental and Clinical Medicine, University of Florence, Viale GB Morgagni 50, Florence 50134, Italy
| | - O Crociani
- Department of Experimental and Clinical Medicine, University of Florence, Viale GB Morgagni 50, Florence 50134, Italy
| | - S Manoli
- Department of Experimental and Clinical Medicine, University of Florence, Viale GB Morgagni 50, Florence 50134, Italy
| | - M D'Amico
- Department of Experimental and Clinical Medicine, University of Florence, Viale GB Morgagni 50, Florence 50134, Italy
- DI.V.A.L Toscana Srl, Via Madonna del Piano 6, Sesto Fiorentino 50019, Italy
| | - M Masselli
- Department of Experimental and Clinical Medicine, University of Florence, Viale GB Morgagni 50, Florence 50134, Italy
| | - J Iorio
- Department of Experimental and Clinical Medicine, University of Florence, Viale GB Morgagni 50, Florence 50134, Italy
| | - M Callea
- Department of Pathology, Pathology Unit, Campus Bio-Medico University, via del Portillo 200, Rome 00128, Italy
| | - D Borzomati
- Department of General Surgery, Campus Bio-Medico University, via del Portillo 200, Rome 00128, Italy
| | - G Nappo
- Department of General Surgery, Campus Bio-Medico University, via del Portillo 200, Rome 00128, Italy
| | - F Bartolozzi
- Casa di Cura Villa Margherita, Viale di Villa Massimo 48, Rome 00161, Italy
| | - D Santini
- Department of Medical Oncology, Campus Bio-Medico University, via del Portillo 200, Rome 00128, Italy
| | - L Bencini
- Department of General Surgery and Surgical Oncology, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla 3, Florence 50134, Italy
| | - M Farsi
- Department of General Surgery and Surgical Oncology, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla 3, Florence 50134, Italy
| | - L Boni
- Clinical Trials Coordinating Center, Azienda Ospedaliero-Universitaria Careggi/Istituto Toscano Tumori, Largo Brambilla 3, Florence 50134, Italy
| | - F Di Costanzo
- Department of Medical Oncology, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla 3, Florence 50134, Italy
| | - A Schwab
- Physiologisches Institut II, University of Münster, Robert-Koch-Str. 27b, Münster D-48149, Germany
| | - A Onetti Muda
- Department of Pathology, Pathology Unit, Campus Bio-Medico University, via del Portillo 200, Rome 00128, Italy
| | - R Coppola
- Department of General Surgery, Campus Bio-Medico University, via del Portillo 200, Rome 00128, Italy
| | - A Arcangeli
- Department of Experimental and Clinical Medicine, University of Florence, Viale GB Morgagni 50, Florence 50134, Italy
| |
Collapse
|